Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 155(5): 1104-18, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24238962

ABSTRACT

Cellular senescence disables proliferation in damaged cells, and it is relevant for cancer and aging. Here, we show that senescence occurs during mammalian embryonic development at multiple locations, including the mesonephros and the endolymphatic sac of the inner ear, which we have analyzed in detail. Mechanistically, senescence in both structures is strictly dependent on p21, but independent of DNA damage, p53, or other cell-cycle inhibitors, and it is regulated by the TGF-ß/SMAD and PI3K/FOXO pathways. Developmentally programmed senescence is followed by macrophage infiltration, clearance of senescent cells, and tissue remodeling. Loss of senescence due to the absence of p21 is partially compensated by apoptosis but still results in detectable developmental abnormalities. Importantly, the mesonephros and endolymphatic sac of human embryos also show evidence of senescence. We conclude that the role of developmentally programmed senescence is to promote tissue remodeling and propose that this is the evolutionary origin of damage-induced senescence.


Subject(s)
Cellular Senescence , Embryonic Development , Endolymphatic Sac/embryology , Mesonephros/embryology , Animals , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Endolymphatic Sac/cytology , Female , Humans , Kidney/embryology , Male , Mesonephros/cytology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
2.
Nature ; 546(7660): 676-680, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28658220

ABSTRACT

Cutaneous melanoma is a type of cancer with an inherent potential for lymph node colonization, which is generally preceded by neolymphangiogenesis. However, sentinel lymph node removal does not necessarily extend the overall survival of patients with melanoma. Moreover, lymphatic vessels collapse and become dysfunctional as melanomas progress. Therefore, it is unclear whether (and how) lymphangiogenesis contributes to visceral metastasis. Soluble and vesicle-associated proteins secreted by tumours and/or their stroma have been proposed to condition pre-metastatic sites in patients with melanoma. Still, the identities and prognostic value of lymphangiogenic mediators remain unclear. Moreover, our understanding of lymphangiogenesis (in melanomas and other tumour types) is limited by the paucity of mouse models for live imaging of distal pre-metastatic niches. Injectable lymphatic tracers have been developed, but their limited diffusion precludes whole-body imaging at visceral sites. Vascular endothelial growth factor receptor 3 (VEGFR3) is an attractive 'lymphoreporter' because its expression is strongly downregulated in normal adult lymphatic endothelial cells, but is activated in pathological situations such as inflammation and cancer. Here, we exploit this inducibility of VEGFR3 to engineer mouse melanoma models for whole-body imaging of metastasis generated by human cells, clinical biopsies or endogenously deregulated oncogenic pathways. This strategy revealed early induction of distal pre-metastatic niches uncoupled from lymphangiogenesis at primary lesions. Analyses of the melanoma secretome and validation in clinical specimens showed that the heparin-binding factor midkine is a systemic inducer of neo-lymphangiogenesis that defines patient prognosis. This role of midkine was linked to a paracrine activation of the mTOR pathway in lymphatic endothelial cells. These data support the use of VEGFR3 reporter mice as a 'MetAlert' discovery platform for drivers and inhibitors of metastasis.


Subject(s)
Cytokines/metabolism , Lymphatic Vessels/metabolism , Neoplasm Metastasis/diagnostic imaging , Neoplasm Metastasis/pathology , Whole Body Imaging/methods , Animals , Disease Models, Animal , Disease Progression , Endothelial Cells/metabolism , Female , Genes, Reporter , Humans , Lymphangiogenesis , Lymphatic Vessels/pathology , Male , Melanoma/diagnostic imaging , Melanoma/metabolism , Melanoma/pathology , Mice , Midkine , Paracrine Communication , Prognosis , Recurrence , Reproducibility of Results , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor Receptor-3/analysis , Vascular Endothelial Growth Factor Receptor-3/metabolism , Xenograft Model Antitumor Assays
3.
PLoS Genet ; 16(12): e1008960, 2020 12.
Article in English | MEDLINE | ID: mdl-33362210

ABSTRACT

Most B cell lymphomas originate from B cells that have germinal center (GC) experience and bear chromosome translocations and numerous point mutations. GC B cells remodel their immunoglobulin (Ig) genes by somatic hypermutation (SHM) and class switch recombination (CSR) in their Ig genes. Activation Induced Deaminase (AID) initiates CSR and SHM by generating U:G mismatches on Ig DNA that can then be processed by Uracyl-N-glycosylase (UNG). AID promotes collateral damage in the form of chromosome translocations and off-target SHM, however, the exact contribution of AID activity to lymphoma generation and progression is not completely understood. Here we show using a conditional knock-in strategy that AID supra-activity alone is not sufficient to generate B cell transformation. In contrast, in the absence of UNG, AID supra-expression increases SHM and promotes lymphoma. Whole exome sequencing revealed that AID heavily contributes to lymphoma SHM, promoting subclonal variability and a wider range of oncogenic variants. Thus, our data provide direct evidence that UNG is a brake to AID-induced intratumoral heterogeneity and evolution of B cell lymphoma.


Subject(s)
Cytidine Deaminase/genetics , Genetic Heterogeneity , Lymphoma, B-Cell/genetics , Uracil-DNA Glycosidase/genetics , Animals , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Clonal Evolution , Cytidine Deaminase/metabolism , Female , Lymphoma, B-Cell/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Uracil-DNA Glycosidase/metabolism
4.
Mov Disord ; 36(4): 895-904, 2021 04.
Article in English | MEDLINE | ID: mdl-33232556

ABSTRACT

BACKGROUND: Recent studies reported abnormal alpha-synuclein deposition in biopsy-accessible sites of the peripheral nervous system in Parkinson's disease (PD). This has considerable implications for clinical diagnosis. Moreover, if deposition occurs early, it may enable tissue diagnosis of prodromal PD. OBJECTIVE: The aim of this study was to develop and test an automated bright-field immunohistochemical assay of cutaneous pathological alpha-synuclein deposition in patients with idiopathic rapid eye movement sleep behavior disorder, PD, and atypical parkinsonism and in control subjects. METHODS: For assay development, postmortem skin biopsies were taken from 28 patients with autopsy-confirmed Lewy body disease and 23 control subjects. Biopsies were stained for pathological alpha-synuclein in automated stainers using a novel dual-immunohistochemical assay for serine 129-phosphorylated alpha-synuclein and pan-neuronal marker protein gene product 9.5. After validation, single 3-mm punch skin biopsies were taken from the cervical 8 paravertebral area from 79 subjects (28 idiopathic rapid eye movement sleep behavior disorder, 20 PD, 10 atypical parkinsonism, and 21 control subjects). Raters blinded to clinical diagnosis assessed the biopsies. RESULTS: The immunohistochemistry assay differentiated alpha-synuclein pathology from nonpathological-appearing alpha-synuclein using combined phosphatase and protease treatments. Among autopsy samples, 26 of 28 Lewy body samples and none of the 23 controls were positive. Among living subjects, punch biopsies were positive in 23 (82%) subjects with idiopathic rapid eye movement sleep behavior disorder, 14 (70%) subjects with PD, 2 (20%) subjects with atypical parkinsonism, and none (0%) of the control subjects. After a 3-year follow-up, eight idiopathic rapid eye movement sleep behavior disorder subjects phenoconverted to defined neurodegenerative syndromes, in accordance with baseline biopsy results. CONCLUSION: Even with a single 3-mm punch biopsy, there is considerable promise for using pathological alpha-synuclein deposition in skin to diagnose both clinical and prodromal PD. © 2020 International Parkinson and Movement Disorder Society.


Subject(s)
Lewy Body Disease , Parkinson Disease , REM Sleep Behavior Disorder , Humans , Skin , alpha-Synuclein
5.
EMBO Rep ; 19(9)2018 09.
Article in English | MEDLINE | ID: mdl-30021836

ABSTRACT

The NAD+-dependent deacetylase SIRT1 can be oncogenic or tumor suppressive depending on the tissue. Little is known about the role of SIRT1 in non-small cell lung carcinoma (NSCLC), one of the deadliest cancers, that is frequently associated with mutated K-RAS Therefore, we investigated the effect of SIRT1 on K-RAS-driven lung carcinogenesis. We report that SIRT1 protein levels are downregulated by oncogenic K-RAS in a MEK and PI3K-dependent manner in mouse embryo fibroblasts (MEFs), and in human lung adenocarcinoma cell lines. Furthermore, Sirt1 overexpression in mice delays the appearance of K-RasG12V-driven lung adenocarcinomas, reducing the number and size of carcinomas at the time of death and extending survival. Consistently, lower levels of SIRT1 are associated with worse prognosis in human NSCLCs. Mechanistically, analysis of mouse Sirt1-Tg pneumocytes, isolated shortly after K-RasG12V activation, reveals that Sirt1 overexpression alters pathways involved in tumor development: proliferation, apoptosis, or extracellular matrix organization. Our work demonstrates a tumor suppressive role of SIRT1 in the development of K-RAS-driven lung adenocarcinomas in mice and humans, suggesting that the SIRT1-K-RAS axis could be a therapeutic target for NSCLCs.


Subject(s)
Adenocarcinoma of Lung/metabolism , Carcinogenesis/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Sirtuin 1/metabolism , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Alveolar Epithelial Cells , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cells, Cultured , Down-Regulation , Fibroblasts/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mitogen-Activated Protein Kinases/metabolism , Molecular Targeted Therapy , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Progression-Free Survival , Proto-Oncogene Proteins p21(ras)/genetics
6.
Nature ; 502(7471): 340-5, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24025773

ABSTRACT

Reprogramming of adult cells to generate induced pluripotent stem cells (iPS cells) has opened new therapeutic opportunities; however, little is known about the possibility of in vivo reprogramming within tissues. Here we show that transitory induction of the four factors Oct4, Sox2, Klf4 and c-Myc in mice results in teratomas emerging from multiple organs, implying that full reprogramming can occur in vivo. Analyses of the stomach, intestine, pancreas and kidney reveal groups of dedifferentiated cells that express the pluripotency marker NANOG, indicative of in situ reprogramming. By bone marrow transplantation, we demonstrate that haematopoietic cells can also be reprogrammed in vivo. Notably, reprogrammable mice present circulating iPS cells in the blood and, at the transcriptome level, these in vivo generated iPS cells are closer to embryonic stem cells (ES cells) than standard in vitro generated iPS cells. Moreover, in vivo iPS cells efficiently contribute to the trophectoderm lineage, suggesting that they achieve a more plastic or primitive state than ES cells. Finally, intraperitoneal injection of in vivo iPS cells generates embryo-like structures that express embryonic and extraembryonic markers. We conclude that reprogramming in vivo is feasible and confers totipotency features absent in standard iPS or ES cells. These discoveries could be relevant for future applications of reprogramming in regenerative medicine.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Teratoma/metabolism , Totipotent Stem Cells/cytology , Animals , Blood Cells/cytology , Blood Cells/metabolism , Cell Dedifferentiation , Cell Separation , Cells, Cultured , Cellular Reprogramming/genetics , Ectoderm/cytology , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Fibroblasts/cytology , Gene Expression Profiling , Induced Pluripotent Stem Cells/metabolism , Intestines/cytology , Kidney/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Organ Specificity , Pancreas/cytology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Stomach/cytology , Teratoma/genetics , Teratoma/pathology , Totipotent Stem Cells/metabolism , Transcriptome/genetics , Trophoblasts/cytology
7.
Hum Mol Genet ; 24(10): 2914-22, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25678551

ABSTRACT

Gastric neuroendocrine tumours (NETs) arise from enterochromaffin-like cells, which are located in oxyntic glands within the stomach. Type I tumours represent 70-80% of gastric NETs and are associated with hypergastrinaemia, chronic atrophic gastritis and achlorhydria. Gastrin is involved in the endocrine regulation of gastric acid production. Most type I gastric NETs are sporadic, have a good prognosis and their genetic basis are unknown. We performed an exome sequencing study in a family with consanguineous parents and 10 children, five of whom were affected by type I gastric NET. Atypical clinical traits included an earlier age of onset (around 30 years), aggressiveness (three had nodal infiltration requiring total gastrectomy and one an adenocarcinoma) and iron-deficiency rather than megaloblastic anaemia. We identified a homozygous missense mutation in the 14th exon of the ATP4A gene (c.2107C>T), which encodes the proton pump responsible for acid secretion by gastric parietal cells. The amino acid p.Arg703Cys is highly conserved across species and originates a change of one of the transmembrane domains that avoids the liberation of protons from cells to stomach. This is consistent with the achlorhydria that was observed in the affected individuals. No germline or somatic mutations in the ATP4A gene were found in sporadic gastric NET patients. Based on the results of this large family, it seems that this atypical form of gastric NET has an earlier age of onset, behaves more aggressively and has atypical clinical traits that differentiated from other studied cases.


Subject(s)
Genetic Predisposition to Disease , H(+)-K(+)-Exchanging ATPase/genetics , Mutation, Missense , Neuroendocrine Tumors/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/genetics , Adolescent , Adult , Aged , Exome , Female , Humans , Male , Middle Aged , Pedigree , Sequence Analysis, DNA , Young Adult
8.
J Pathol ; 239(2): 206-17, 2016 06.
Article in English | MEDLINE | ID: mdl-27174785

ABSTRACT

The Noonan syndrome (NS) is an autosomal dominant genetic disorder characterized by short stature, craniofacial dysmorphism, and congenital heart defects. A significant proportion of NS patients may also develop myeloproliferative disorders (MPDs), including juvenile myelomonocytic leukaemia (JMML). Surprisingly, scarce information is available in relation to other tumour types in these patients. We have previously developed and characterized a knock-in mouse model that carries one of the most frequent KRAS-NS-related mutations, the K-Ras(V14I) substitution, which recapitulates most of the alterations described in NS patients, including MPDs. The K-Ras(V14I) mutation is a mild activating K-Ras protein; thus, we have used this model to study tumour susceptibility in comparison with mice expressing the classical K-Ras(G12V) oncogene. Interestingly, our studies have shown that these mice display a generalized tumour predisposition and not just MPDs. In fact, we have observed that the K-Ras(V14I) mutation is capable of cooperating with the p16Ink4a/p19Arf and Trp53 tumour suppressors, as well as with other risk factors such as pancreatitis, thereby leading to a higher cancer incidence. In conclusion, our results illustrate that the K-Ras(V14I) activating protein is able to induce cancer, although at a much lower level than the classical K-Ras(G12V) oncogene, and that it can be significantly modulated by both genetic and non-genetic events. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Heart Defects, Congenital/genetics , Lung Neoplasms/genetics , Neoplasms/genetics , Noonan Syndrome/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Alleles , Amino Acid Substitution , Animals , Disease Models, Animal , Disease Susceptibility , Female , Genetic Carrier Screening , Heart Defects, Congenital/pathology , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mutation , Neoplasms/pathology , Noonan Syndrome/pathology , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism
9.
Proc Natl Acad Sci U S A ; 111(46): 16395-400, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25359213

ABSTRACT

Noonan syndrome (NS) is an autosomal dominant genetic disorder characterized by short stature, craniofacial dysmorphism, and congenital heart defects. NS also is associated with a risk for developing myeloproliferative disorders (MPD), including juvenile myelomonocytic leukemia (JMML). Mutations responsible for NS occur in at least 11 different loci including KRAS. Here we describe a mouse model for NS induced by K-Ras(V14I), a recurrent KRAS mutation in NS patients. K-Ras(V14I)-mutant mice displayed multiple NS-associated developmental defects such as growth delay, craniofacial dysmorphia, cardiac defects, and hematologic abnormalities including a severe form of MPD that resembles human JMML. Homozygous animals had perinatal lethality whose penetrance varied with genetic background. Exposure of pregnant mothers to a MEK inhibitor rescued perinatal lethality and prevented craniofacial dysmorphia and cardiac defects. However, Mek inhibition was not sufficient to correct these defects when mice were treated after weaning. Interestingly, Mek inhibition did not correct the neoplastic MPD characteristic of these mutant mice, regardless of the timing at which the mice were treated, thus suggesting that MPD is driven by additional signaling pathways. These genetically engineered K-Ras(V14I)-mutant mice offer an experimental tool for studying the molecular mechanisms underlying the clinical manifestations of NS. Perhaps more importantly, they should be useful as a preclinical model to test new therapies aimed at preventing or ameliorating those deficits associated with this syndrome.


Subject(s)
Disease Models, Animal , Genes, ras , Mice, Mutant Strains , Mutation, Missense , Noonan Syndrome/genetics , Point Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Abnormalities, Multiple/embryology , Abnormalities, Multiple/genetics , Abnormalities, Multiple/prevention & control , Alleles , Amino Acid Substitution , Animals , Body Size/genetics , Cell Lineage , Crosses, Genetic , Dwarfism/genetics , Epistasis, Genetic , Face/abnormalities , Female , Genes, Dominant , Genotype , Heart Defects, Congenital/genetics , Hematopoiesis/genetics , Leukemia, Myelomonocytic, Juvenile/genetics , MAP Kinase Kinase Kinases/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains/genetics , Myeloproliferative Disorders/genetics , Neoplastic Syndromes, Hereditary/embryology , Neoplastic Syndromes, Hereditary/genetics , Phenotype , Pregnancy , Prenatal Exposure Delayed Effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/physiology , Radiation Chimera , Signal Transduction/drug effects
10.
Gut ; 65(3): 476-86, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25596178

ABSTRACT

BACKGROUND AND AIMS: Gata6 is required to complete and maintain acinar differentiation in the mouse pancreas. Pancreas-specific Gata6 ablation during development causes extensive and persistent acinar-ductal metaplasia, which is considered an initial step of mutant KRas-driven carcinogenesis. Therefore, the Gata6-null pancreas might represent a tumour-prone environment. We investigated whether Gata6 plays a role during pancreatic tumorigenesis. DESIGN: We analysed genetically engineered mouse models and human pancreatic ductal adenocarcinoma (PDAC) cell lines, using a combination of histopathological studies, genome-wide expression and chromatin immunoprecipitation experiments to understand the role of Gata6 in the initiation and progression of KRas(G12V)-driven tumours RESULTS: We show that Gata6 maintains the acinar differentiation programme, both directly and indirectly, and it concomitantly suppresses ectopic programmes in the pancreas. Gata6 ablation renders acinar cells more sensitive to KRas(G12V), thereby accelerating tumour development. Gata6 expression is spontaneously lost in a mouse model of KRas(G12V)-driven PDAC, in association with altered cell differentiation. Using a combination of ChIP-Seq and RNA-Seq, we show that Gata6 exerts its tumour-suppressive effect through the promotion of cell differentiation, the suppression of inflammatory pathways, and the direct repression of cancer-related pathways. Among them is the epidermal growth factor receptor (EGFR) pathway, the activity of which is upregulated in the normal and preneoplastic Gata6-null pancreas. Accordingly, GATA6-silencing in human PDAC cells leads to an upregulation of EGFR. CONCLUSIONS: We propose that, in the pancreas, Gata6 acts as a tumour suppressor by enforcing acinar cell differentiation, by directly and indirectly repressing ectopic differentiation programmes, and by regulating crucial cancer-related gene expression pathways.


Subject(s)
Acinar Cells/physiology , Biomarkers, Tumor/metabolism , Carcinogenesis/metabolism , Carcinoma, Pancreatic Ductal/metabolism , GATA6 Transcription Factor/metabolism , Pancreatic Neoplasms/metabolism , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Differentiation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Transgenic , Mutation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics
11.
EMBO J ; 31(9): 2076-89, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22415365

ABSTRACT

Cohesin is a protein complex originally identified for its role in sister chromatid cohesion, although increasing evidence portrays it also as a major organizer of interphase chromatin. Vertebrate cohesin consists of Smc1, Smc3, Rad21/Scc1 and either stromal antigen 1 (SA1) or SA2. To explore the functional specificity of these two versions of cohesin and their relevance for embryonic development and cancer, we generated a mouse model deficient for SA1. Complete ablation of SA1 results in embryonic lethality, while heterozygous animals have shorter lifespan and earlier onset of tumourigenesis. SA1-null mouse embryonic fibroblasts show decreased proliferation and increased aneuploidy as a result of chromosome segregation defects. These defects are not caused by impaired centromeric cohesion, which depends on cohesin-SA2. Instead, they arise from defective telomere replication, which requires cohesion mediated specifically by cohesin-SA1. We propose a novel mechanism for aneuploidy generation that involves impaired telomere replication upon loss of cohesin-SA1, with clear implications in tumourigenesis.


Subject(s)
Aneuploidy , Cell Cycle Proteins/deficiency , Chromosomal Proteins, Non-Histone/deficiency , Protein Subunits/deficiency , Telomere/metabolism , Animals , Carcinogens , Cell Cycle Proteins/genetics , Cell Line , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation , Diethylnitrosamine , Fibrosarcoma/chemically induced , Fibrosarcoma/genetics , Fibrosarcoma/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Methylcholanthrene , Mice , Mice, Knockout , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Protein Subunits/genetics , Sister Chromatid Exchange , Cohesins
12.
EMBO J ; 31(11): 2498-510, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22505032

ABSTRACT

Cyclin-dependent kinase (Cdk)7, the catalytic subunit of the Cdk-activating kinase (CAK) complex has been implicated in the control of cell cycle progression and of RNA polymerase II (RNA pol II)-mediated transcription. Genetic inactivation of the Cdk7 locus revealed that whereas Cdk7 is completely dispensable for global transcription, is essential for the cell cycle via phosphorylation of Cdk1 and Cdk2. In vivo, Cdk7 is also indispensable for cell proliferation except during the initial stages of embryonic development. Interestingly, widespread elimination of Cdk7 in adult tissues with low proliferative indexes had no phenotypic consequences. However, ablation of conditional Cdk7 alleles in tissues with elevated cellular turnover led to the efficient repopulation of these tissues with Cdk7-expressing cells most likely derived from adult stem cells that may have escaped the inactivation of their targeted Cdk7 alleles. This process, a physiological attempt to maintain tissue homeostasis, led to the attrition of adult stem cell pools and to the appearance of age-related phenotypes, including telomere shortening and early death.


Subject(s)
Adult Stem Cells/physiology , Aging, Premature/genetics , Cell Cycle Checkpoints/physiology , Cyclin-Dependent Kinases/physiology , Animals , Cell Cycle Checkpoints/genetics , Cell Proliferation , Cyclin-Dependent Kinases/genetics , Embryonic Development/physiology , Female , Homeostasis/physiology , Mice , Telomere Shortening/physiology
13.
Blood ; 124(15): 2380-90, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25157181

ABSTRACT

Cdk4 and Cdk6 are related protein kinases that bind d-type cyclins and regulate cell-cycle progression. Cdk4/6 inhibitors are currently being used in advanced clinical trials and show great promise against many types of tumors. Cdk4 and Cdk6 are inhibited by INK4 proteins, which exert tumor-suppressing functions. To test the significance of this inhibitory mechanism, we generated knock-in mice that express a Cdk6 mutant (Cdk6 R31C) insensitive to INK4-mediated inhibition. Cdk6(R/R) mice display altered development of the hematopoietic system without enhanced tumor susceptibility, either in the presence or absence of p53. Unexpectedly, Cdk6 R31C impairs the potential of hematopoietic progenitors to repopulate upon adoptive transfer or after 5-fluorouracil-induced damage. The defects are overcome by eliminating sensitivity of cells to INK4 inhibitors by introducing the INK4-insensitive Cdk4 R24C allele, and INK4-resistant mice are more susceptible to hematopoietic and endocrine tumors. In BCR-ABL-transformed hematopoietic cells, Cdk6 R31C causes increased binding of p16(INK4a) to wild-type Cdk4, whereas cells harboring Cdk4 R24C and Cdk6 R31C are fully insensitive to INK4 inhibitors, resulting in accelerated disease onset. Our observations reveal that Cdk4 and Cdk6 cooperate in hematopoietic tumor development and suggest a role for Cdk6 in sequestering INK4 proteins away from Cdk4.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Alleles , Animals , Cell Death , Cell Line, Transformed , Cell Proliferation , Cyclin-Dependent Kinase 6/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Ontology , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Mice , Mutant Proteins/metabolism
14.
Blood ; 124(2): 229-39, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24850757

ABSTRACT

microRNAs are a class of regulators of gene expression that have been shown critical for a great number of biological processes; however, little is known of their role in germinal center (GC) B cells. Although the GC reaction is crucial to ensure a competent immune response, GC B cells are also the origin of most human lymphomas, presumably due to bystander effects of the immunoglobulin gene remodeling that takes place at these sites. Here we report that miR-217 is specifically upregulated in GC B cells. Gain- and loss-of-function mouse models reveal that miR-217 is a positive modulator of the GC response that increases the generation of class-switched antibodies and the frequency of somatic hypermutation. We find that miR-217 down-regulates the expression of a DNA damage response and repair gene network and in turn stabilizes Bcl-6 expression in GC B cells. Importantly, miR-217 overexpression also promotes mature B-cell lymphomagenesis; this is physiologically relevant as we find that miR-217 is overexpressed in aggressive human B-cell lymphomas. Therefore, miR-217 provides a novel molecular link between the normal GC response and B-cell transformation.


Subject(s)
Germinal Center/physiology , MicroRNAs/physiology , Oncogenes/physiology , Animals , B-Lymphocytes/pathology , B-Lymphocytes/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cells, Cultured , DNA Damage/genetics , DNA Repair/genetics , Gene Regulatory Networks , Lymphoma/genetics , Lymphoma/metabolism , Mice , Mice, Transgenic , Microarray Analysis , Proto-Oncogene Proteins c-bcl-6/genetics
15.
Proc Natl Acad Sci U S A ; 110(34): 13791-6, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23918357

ABSTRACT

Antibody cancer therapies rely on systemically accessible targets and suitable antibodies that exert a functional activity or deliver a payload to the tumor site. Here, we present proof-of-principle of in vivo selection of human antibodies in tumor-bearing mice that identified a tumor-specific antibody able to deliver a payload and unveils the target antigen. By using an ex vivo enrichment process against freshly disaggregated tumors to purge the repertoire, in combination with in vivo biopanning at optimized phage circulation time, we have identified a human domain antibody capable of mediating selective localization of phage to human prostate cancer xenografts. Affinity chromatography followed by mass spectrometry analysis showed that the antibody recognizes the proteasome activator complex PA28. The specificity of soluble antibody was confirmed by demonstrating its binding to the active human PA28αß complex. Whereas systemically administered control phage was confined in the lumen of blood vessels of both normal tissues and tumors, the selected phage spread from tumor vessels into the perivascular tumor parenchyma. In these areas, the selected phage partially colocalized with PA28 complex. Furthermore, we found that the expression of the α subunit of PA28 [proteasome activator complex subunit 1 (PSME1)] is elevated in primary and metastatic human prostate cancer and used anti-PSME1 antibodies to show that PSME1 is an accessible marker in mouse xenograft tumors. These results support the use of PA28 as a tumor marker and a potential target for therapeutic intervention in prostate cancer.


Subject(s)
Antibodies, Neoplasm/immunology , Biomarkers, Tumor/immunology , Immunotherapy/methods , Muscle Proteins/metabolism , Prostatic Neoplasms/immunology , Proteasome Endopeptidase Complex/metabolism , Animals , Antibodies, Neoplasm/metabolism , Antibody Specificity , Blotting, Western , Cell Surface Display Techniques , Chromatography, Affinity , Chromatography, Liquid , Drug Delivery Systems/methods , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Immunoprecipitation , Male , Mice , Mice, Nude , Prostatic Neoplasms/therapy , Statistics, Nonparametric , Tandem Mass Spectrometry
16.
Gut ; 64(12): 1921-35, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25841238

ABSTRACT

OBJECTIVES: The tumour stroma/microenvironment not only provides structural support for tumour development, but more importantly it provides cues to cancer stem cells (CSCs) that regulate their self-renewal and metastatic potential. This is certainly true for pancreatic ductal adenocarcinomas (PDAC), where tumour-associated fibroblasts, pancreatic stellate cells and immune cells create an abundant paracrine niche for CSCs via microenvironment-secreted factors. Thus understanding the role that tumour stroma cells play in PDAC development and CSC biology is of utmost importance. DESIGN: Microarray analyses, tumour microarray immunohistochemical assays, in vitro co-culture experiments, recombinant protein treatment approaches and in vivo intervention studies were performed to understand the role that the immunomodulatory cationic antimicrobial peptide 18/LL-37 (hCAP-18/LL-37) plays in PDAC biology. RESULTS: We found that hCAP-18/LL-37 was strongly expressed in the stroma of advanced primary and secondary PDAC tumours and is secreted by immune cells of the stroma (eg, tumour-associated macrophages) in response to tumour growth factor-ß1 and particularly CSC-secreted Nodal/ActivinA. Treatment of pancreatic CSCs with recombinant LL-37 increased pluripotency-associated gene expression, self-renewal, invasion and tumourigenicity via formyl peptide receptor 2 (FPR2)- and P2X purinoceptor 7 receptor (P2X7R)-dependent mechanisms, which could be reversed by inhibiting these receptors. Importantly, in a genetically engineered mouse model of K-Ras-driven pancreatic tumourigenesis, we also showed that tumour formation was inhibited by either reconstituting these mice with bone marrow from cathelicidin-related antimicrobial peptide (ie, murine homologue of hCAP-18/LL-37) knockout mice or by pharmacologically inhibiting FPR2 and P2X7R. CONCLUSIONS: Thus, hCAP-18/LL-37 represents a previously unrecognised PDAC microenvironment factor that plays a critical role in pancreatic CSC-mediated tumourigenesis.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Activins/metabolism , Animals , Antimicrobial Cationic Peptides/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Pancreatic Ductal/genetics , Cell Self Renewal/drug effects , Gene Expression/drug effects , Humans , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplastic Stem Cells/drug effects , Pancreatic Neoplasms/genetics , Protein Array Analysis , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Formyl Peptide/metabolism , Receptors, Purinergic P2X7/metabolism , Signal Transduction/drug effects , Tissue Array Analysis , Transforming Growth Factor beta1/pharmacology , Cathelicidins
17.
Gastroenterology ; 147(5): 1119-33.e4, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25127677

ABSTRACT

BACKGROUND & AIMS: Although smoking is a leading risk factor for pancreatic ductal adenocarcinoma (PDAC), little is known about the mechanisms by which smoking promotes initiation or progression of PDAC. METHODS: We studied the effects of nicotine administration on pancreatic cancer development in Kras(+/LSLG12Vgeo);Elas-tTA/tetO-Cre (Ela-KRAS) mice, Kras(+/LSLG12D);Trp53+/LSLR172H;Pdx-1-Cre (KPC) mice (which express constitutively active forms of KRAS), and C57/B6 mice. Mice were given nicotine for up to 86 weeks to produce blood levels comparable with those of intermediate smokers. Pancreatic tissues were collected and analyzed by immunohistochemistry and reverse transcriptase polymerase chain reaction; cells were isolated and assayed for colony and sphere formation and gene expression. The effects of nicotine were also evaluated in primary pancreatic acinar cells isolated from wild-type, nAChR7a(-/-), Trp53(-/-), and Gata6(-/-);Trp53(-/-) mice. We also analyzed primary PDAC cells that overexpressed GATA6 from lentiviral expression vectors. RESULTS: Administration of nicotine accelerated transformation of pancreatic cells and tumor formation in Ela-KRAS and KPC mice. Nicotine induced dedifferentiation of acinar cells by activating AKT-ERK-MYC signaling; this led to inhibition of Gata6 promoter activity, loss of GATA6 protein, and subsequent loss of acinar differentiation and hyperactivation of oncogenic KRAS. Nicotine also promoted aggressiveness of established tumors as well as the epithelial-mesenchymal transition, increasing numbers of circulating cancer cells and their dissemination to the liver, compared with mice not exposed to nicotine. Nicotine induced pancreatic cells to acquire gene expression patterns and functional characteristics of cancer stem cells. These effects were markedly attenuated in K-Ras(+/LSL-G12D);Trp53(+/LSLR172H);Pdx-1-Cre mice given metformin. Metformin prevented nicotine-induced pancreatic carcinogenesis and tumor growth by up-regulating GATA6 and promoting differentiation toward an acinar cell program. CONCLUSIONS: In mice, nicotine promotes pancreatic carcinogenesis and tumor development via down-regulation of Gata6 to induce acinar cell dedifferentiation.


Subject(s)
Acinar Cells/drug effects , Carcinoma, Pancreatic Ductal/chemically induced , Cell Dedifferentiation/drug effects , GATA6 Transcription Factor/metabolism , Nicotine/toxicity , Nicotinic Agonists/toxicity , Pancreas/drug effects , Pancreatic Neoplasms/chemically induced , Proto-Oncogene Proteins p21(ras)/metabolism , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/prevention & control , Carcinoma, Pancreatic Ductal/secondary , Cell Line, Tumor , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial-Mesenchymal Transition/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , GATA6 Transcription Factor/deficiency , GATA6 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Metformin/pharmacology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Mutation , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/prevention & control , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/deficiency , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction/drug effects , Time Factors , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics
18.
Cancer Cell ; 11(3): 291-302, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17349585

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA), one of the deadliest human cancers, often involves somatic activation of K-Ras oncogenes. We report that selective expression of an endogenous K-Ras(G12V) oncogene in embryonic cells of acinar/centroacinar lineage results in pancreatic intraepithelial neoplasias (PanINs) and invasive PDA, suggesting that PDA originates by differentiation of acinar/centroacinar cells or their precursors into ductal-like cells. Surprisingly, adult mice become refractory to K-Ras(G12V)-induced PanINs and PDA. However, if these mice are challenged with a mild form of chronic pancreatitis, they develop the full spectrum of PanINs and invasive PDA. These observations suggest that, during adulthood, PDA stems from a combination of genetic (e.g., somatic K-Ras mutations) and nongenetic (e.g., tissue damage) events.


Subject(s)
Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/pathology , Genes, ras , Pancreatic Neoplasms/pathology , Pancreatitis, Chronic/pathology , Animals , Carcinoma in Situ/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Cell Lineage , Cell Transformation, Neoplastic , Ceruletide , Doxycycline/pharmacology , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Mice , Mice, Mutant Strains , Mutation , Neoplasm Invasiveness , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Pancreatitis, Chronic/chemically induced , Signal Transduction
19.
Nature ; 460(7259): 1136-9, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19668188

ABSTRACT

The mechanisms involved in the reprogramming of differentiated cells into induced pluripotent stem (iPS) cells by the three transcription factors Oct4 (also known as Pou5f1), Klf4 and Sox2 remain poorly understood. The Ink4/Arf locus comprises the Cdkn2a-Cdkn2b genes encoding three potent tumour suppressors, namely p16(Ink4a), p19(Arf) and p15(Ink4b), which are basally expressed in differentiated cells and upregulated by aberrant mitogenic signals. Here we show that the locus is completely silenced in iPS cells, as well as in embryonic stem (ES) cells, acquiring the epigenetic marks of a bivalent chromatin domain, and retaining the ability to be reactivated after differentiation. Cell culture conditions during reprogramming enhance the expression of the Ink4/Arf locus, further highlighting the importance of silencing the locus to allow proliferation and reprogramming. Indeed, the three factors together repress the Ink4/Arf locus soon after their expression and concomitant with the appearance of the first molecular markers of 'stemness'. This downregulation also occurs in cells carrying the oncoprotein large-T, which functionally inactivates the pathways regulated by the Ink4/Arf locus, thus indicating that the silencing of the locus is intrinsic to reprogramming and not the result of a selective process. Genetic inhibition of the Ink4/Arf locus has a profound positive effect on the efficiency of iPS cell generation, increasing both the kinetics of reprogramming and the number of emerging iPS cell colonies. In murine cells, Arf, rather than Ink4a, is the main barrier to reprogramming by activation of p53 (encoded by Trp53) and p21 (encoded by Cdkn1a); whereas, in human fibroblasts, INK4a is more important than ARF. Furthermore, organismal ageing upregulates the Ink4/Arf locus and, accordingly, reprogramming is less efficient in cells from old organisms, but this defect can be rescued by inhibiting the locus with a short hairpin RNA. All together, we conclude that the silencing of Ink4/Arf locus is rate-limiting for reprogramming, and its transient inhibition may significantly improve the generation of iPS cells.


Subject(s)
Cellular Reprogramming/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Aging/physiology , Animals , Cell Count , Cell Differentiation , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/genetics , Embryonic Stem Cells/cytology , Epigenesis, Genetic , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Silencing , Humans , Keratinocytes , Kinetics , Kruppel-Like Factor 4 , Mice , Mice, Inbred C57BL
20.
Gut ; 63(4): 647-55, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23598351

ABSTRACT

OBJECTIVES: Nr5a2 participates in biliary acid metabolism and is a major regulator of the pancreatic exocrine programme. Single nucleotide polymorphisms in the vicinity of NR5A2 are associated with the risk of pancreatic ductal adenocarcinoma (PDAC). AIMS: To determine the role of Nr5a2 in pancreatic homeostasis, damage-induced regeneration and mutant KRas-driven pancreatic tumourigenesis. DESIGN: Nr5a2+/- and KRas(G12V);Ptf1a-Cre;Nr5a2+/- mice were used to investigate whether a full dose of Nr5a2 is required for normal pancreas development, recovery from caerulein-induced pancreatitis, and protection from tumour development. RESULTS: Adult Nr5a2+/- mice did not display histological abnormalities in the pancreas but showed a more severe acute pancreatitis, increased acino-ductal metaplasia and impaired recovery from damage. This was accompanied by increased myeloid cell infiltration and proinflammatory cytokine gene expression, and hyperactivation of nuclear factor κb and signal transducer and activator of transcription 3 signalling pathways. Induction of multiple episodes of acute pancreatitis was associated with more severe damage and delayed regeneration. Inactivation of one Nr5a2 allele selectively in pancreatic epithelial cells was sufficient to cause impaired recovery from pancreatitis. In comparison with Nr5a2+/+ mice, KRas(G12V);Ptf1a(Cre/+);Nr5a2+/- mice showed a non-statistically significant increase in the area affected by preneoplastic lesions. However, a single episode of acute pancreatitis cooperated with loss of one Nr5a2 allele to accelerate KRas(G12V)-driven development of preneoplastic lesions. CONCLUSIONS: A full Nr5a2 dose is required to restore pancreatic homeostasis upon damage and to suppress the KRas(G12V)-driven mouse pancreatic intraepithelial neoplasia progression, indicating that Nr5a2 is a novel pancreatic tumour suppressor. Nr5a2 could contribute to PDAC through a role in the recovery from pancreatitis-induced damage.


Subject(s)
Pancreatic Neoplasms/genetics , Pancreatitis/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Blotting, Western , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/physiopathology , Ceruletide/pharmacology , Heterozygote , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , NF-kappa B/physiology , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/physiopathology , Pancreatitis/chemically induced , Pancreatitis/physiopathology , Polymerase Chain Reaction , Proto-Oncogene Proteins p21(ras)/physiology , Receptors, Cytoplasmic and Nuclear/physiology , STAT3 Transcription Factor/physiology , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL